Search for


TEXT SIZE

search for



CrossRef (0)
Arctigenin Inhibits the Viability of Estrogen Receptor Negative Breast Cancer Cells by Inducing Apoptosis
Biomed Sci Letters 2024;30:209-218
Published online December 31, 2024;  https://doi.org/10.15616/BSL.2024.30.4.209
© 2024 The Korean Society For Biomedical Laboratory Sciences.

Tae Hyun Yoo1,*, Hyun Jun Woo2,*, and Sa-Hyun Kim3,4,†,*

1Department of Clinical Laboratory Science, Dongnam Health University, Suwon 16328, Korea
2Department of Clinical Laboratory Science, Semyung University, Jaecheon 27136, Korea
3Departmentof Biotechnology, Kyungpook National University, Daegu 41566, Korea
4BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Korea
Correspondence to: Sa-Hyun Kim
Department of Biotechnology, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Korea
Tel: +82-53-950-5374, Fax: +82-50-4411-9604
E-mail: kimsh78@knu.ac.kr
ORCID: https://orcid.org/0000-0003-3910-4838

*Associate professor.
Received October 21, 2024; Revised October 26, 2024; Accepted November 8, 2024.
This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/3.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
 Abstract
Objectives: Arctigenin, a lignan extracted from the greater burdock has been traditionally used as a medicinal herb. Breast cancer researchers use three markers to classify the type of breast cancer: estrogen receptor (ER), human epidermal growth factor receptor 2 (HER2), and progesterone receptor (PR). This study investigated the anticancer effects of arctigenin and its mechanisms of action in three breast cancer cell lines: MCF-7 (ER+/HER2+/PR–), SK-BR-3 (ER–/HER2+/PR–), and MDA-MB-231 (ER–/HER2–/PR–).
Methods: The anticancer efficacy was assessed using the WST cell viability assay, while the mechanisms of apoptosis induction were analyzed through Annexin V-FITC/PI staining and Western blotting.
Results: In our results, arctigenin at a concentration of less than 50 uM did not inhibit the growth of ER-positive MCF-7 cells. In contrast, arctigenin at concentrations ranging from 0.39 to 50 uM inhibited the development of ER-negative cells, SK-BR-3 and MDA-MB-231. Western blot analysis and annexin V-FITC/PI analysis revealed that apoptosis was involved in the death of arctigenin-treated cancer cells. Specifically, it was observed that increased levels of cleaved PARP, cleaved caspase-3, and cleaved caspase-9, along with decreased levels of XIAP and Survivin, and an increased Bax/Bcl-2 ratio in arctigenin-treated SK-BR-3 and MDA-MB-231 cells.
Conclusion: These results indicated that arctigenin induced cell death by activating apoptosis in ER–/HER2+ SK-BR-3 and ER–/HER2– MDA-MB-231 breast cancer cells. Consequently, these results support the potential use of arctigenin as a therapeutic agent for ER– breast cancer treatment.
Keywords : Apoptosis, Arctigenin, Breast neoplasms, MDA-MB-231, SK-BR-3
INTRODUCTION

Arctigenin, a phytochemical derived from the Asteraceae family, specifically from Arctium lappa (commonly known as burdock), is widely utilized in traditional medicine across Asia to address infections such as sore throat, boils, rashes, and various skin disorders (1). The roots of Arctium lappa are also consumed globally as a vegetable. Arctigenin has been demonstrated to exhibit a range of biological activities, including antiviral, antimicrobial, anti-inflammatory, and immunomodulatory effects (2-4). Furthermore, it has shown significant anticancer activity against various human cancer cell lines, including those associated with gastric, glioblastoma, bladder, lung, and breast cancers (5-9).

Apoptosis, a form of programmed cell death, is crucial for maintaining cellular homeostasis and is involved in various physiological processes including cell turnover, immune system functionality, and embryonic development. Dysregulation of apoptosis can lead to numerous diseases, including developmental disorders, autoimmune diseases, neurodegeneration, and cancer (10). A complex network of proteins regulates apoptosis and can be classified into two main pathways: the intrinsic (mitochondrial) pathway and the extrinsic (death receptor) pathway. Apoptosis is tightly regulated due to its irreversible nature once initiated.

The control of apoptosis in the intrinsic pathway is mediated by the Bcl-2 protein family, which is categorized into three classes: anti-apoptotic proteins (e.g., Bcl-2, Bcl-xL, Mcl-1), pro-apoptotic proteins (e.g., Bax, Bak), and BH3-only proteins (e.g., Bad, Bik, Bid, Bim, Bok). The Bax/Bcl-2 ratio serves as a critical determinant of a cell’s susceptibility to apoptosis, with lower ratios potentially conferring resistance to apoptosis. This ratio also influences tumor progression and aggressiveness (11).

Inhibitors of apoptosis proteins (IAPs) are classified into three groups: I (e.g., X-linked IAP, XIAP), II, and III. XIAP, a prominent member of class I IAPs, is a potent inhibitor of caspase activity, specifically binding to and inhibiting caspases 3, 7, and 9. Survivin, a member of the smallest class III IAPs, inhibits caspase 9 activity by interacting with upstream regulators involved in mitochondrial-dependent apoptosis (12,13).

Breast cancer is the most prevalent cancer and the second leading cause of cancer-related deaths among women globally, following lung cancer (14,15). It is a complex and heterogeneous disease, typically classified based on receptor status into three categories: estrogen receptor-positive (ER+), human epidermal growth factor receptor 2-overexpressing (HER2+), or triple-negative breast cancer (TNBC), characterized by the absence of estrogen, HER2, and progesterone receptors (16). Receptor status is crucial for determining treatment strategies, such as tamoxifen for ER+ cancers or trastuzumab for HER2+ cancers. While ER+ cancers, which are dependent on estrogen for growth, generally have a better prognosis with estrogen-reducing therapies, HER2+ cancers have seen significant improvement in prognosis due to targeted treatments like trastuzumab (17). In contrast, TNBCs, lacking targeted therapies, have a poorer prognosis and are often associated with drug resistance and metastatic spread, highlighting the need for new therapeutic agents.

Thus, this study evaluates the cytotoxicity of arctigenin against ER–/HER2+/PR– type (SK-BR-3) or TNBC, that is to say, ER–/HER2–/PR– type (MDA-MB-231) cells and investigates its effects on apoptosis in these cancer cells.

MATERIALS AND METHODS

1. Materials

Actigenin used in this study was purchased from Sigma-Aldrich. Actigenin was dissolved using dimethyl sulfoxide and then diluted with experimental media in the experiment.

2. Cell culture

The AGS gastric adenocarcinoma cells (American type cell collection CRT-1739) and breast cancer cell lines MCF-7, SK-BR-3, and MDA-MB-231 were obtained from the Korean Cell Line Bank. AGS cells were cultured in DMEM supplemented with 10% fetal bovine serum and streptomycin-penicillin (100 μg/mL and 100 IU/mL). MCF-7, SK-BR-3, MDA-MB-231, and cells were maintained in RPMI 1,640 medium with 10% fetal bovine serum and streptomycin-penicillin (100 μg/mL and 100 IU/mL). All cell lines were incubated at 37°C in a humidified atmosphere with 5% CO2.

3. WST cell viability assay

Cells were seeded into 96-well plates at a density of 1 × 104 cells per well. After 24 hours, the cells were treated with various concentrations of arctigenin (0–50 μM) and incubated for an additional 24 hours. Cell viability was assessed using the EZ-Cytox cell viability assay kit (DoGenBio) according to the manufacturer’s instructions. Ten microliters of WST solution were added to the culture medium, followed by a 2-hour incubation in the CO2 incubator. The Absorbance was measured at 450 nm using a spectrophotometer.

4. Annexin V-FITC and PI staining

Apoptosis was detected using the Annexin V-FITC Apoptosis Detection Kit I according to the manufacturer’s protocol. Cells were trypsinized, washed twice with cold phosphate beffered saline (PBS), and centrifuged at 1,763 g for 5 minutes. The pellet was resuspended in 500 μL of binding buffer at a concentration of 5 × 105 cells/mL, and 100 μL of the cell suspension was transferred to a new 5 mL tube. Then, 5 μL of Annexin V-FITC and 5 μL of propidium iodide (PI) were added. The mixture was incubated for 10 minutes at 37°C in the dark. Following incubation, 400 μL of binding buffer was added, and the samples were analyzed by flow cytometry.

5. Western blotting

The cells were washed with PBS and lysed at 4°C with a buffer containing 1% Triton X-100, a protease inhibitor cocktail, and PBS. The lysates were centrifuged at 7,605 g for 10 minutes at 4°C, and the supernatants were collected. Protein concentrations were determined using the Lowry protein assay. Proteins were separated by SDS-PAGE for 90 minutes at 120 volts and transferred to a nitrocellulose membrane for 90 minutes at 400 mA. The membrane was incubated overnight at 4°C with primary antibodies at optimal concentrations, followed by a 1-hour incubation with the appropriate secondary antibodies at room temperature. The antibodies used are listed in Table 1, with GAPDH serving as an internal control. Immunoreactive proteins were visualized using enhanced chemiluminescence solution.

List of antibodies used in the present study

Name of antibody Species Dilution ratio Molecular weight (kDa) Supplier (Catalog No.)
PARP Rabbit 1:1,000 89, 116 Cell signaling (9496)
Cleaved caspase 3 Rabbit 1:3,000 17, 19, 32 Cell signaling (9661)
Cleaved caspase 8 Rabbit 1:3,000 18, 41, 43 Abcam (ab227430)
Cleaved caspase 9 Rabbit 1:3,000 35, 37, 47 Cell signaling (9502)
Bax Rabbit 1:3,000 20 Cell signaling (2772)
Bcl2 Mouse 1:1,000 26 Cell signaling (4223)
XIAP Rabbit 1:3,000 53 Cell signaling (2045)
Survivin Rabbit 1:1,000 16 Cell signaling (2808)
GAPDH Mouse 1:1,000 37 Santa Cruz (sc-47724)


6. Statistical analysis

Data in the bar graphs are presented as mean ± standard error of the mean. Statistical analyses were conducted using GraphPad Prism 5.02 software (GraphPad Software). Data were analyzed by unpaired Student’s t-test and P < 0.05 was considered to be statistically significant (*P < 0.05, **P < 0.01 and ***P < 0.001).

RESULTS

1. Effect of arctigenin on the viability of cancer cells

To investigate its potential impact on gastric cancer, we assessed the effect of arctigenin on AGS cells, a gastric adenocarcinoma cell line. AGS cells were treated with arctigenin at concentrations ranging from 0.39 μM to 50 μM for 24 hours, and cell viability was measured using the WST assay. No significant changes in cell viability were observed in AGS cells (Fig. 1A). In addition, we evaluated the effect of arctigenin on the viability of various breast cancer cell lines: MCF-7 (ER+/HER2–/PR–), SK-BR-3 (ER–/HER2+/PR–), and MDA-MB-231 (ER–/HER2–/PR–) cells. Cells were treated with arctigenin at concentrations between 0.39 μM and 50 μM for 24 hours, followed by the WST assay. ER+ MCF-7 cells exhibited almost no significant effects at lower concentrations than 50 μM of arctigenin (Fig. 1B). In contrast, the viability of ER– SK-BR-3 and ER– MDA-MB-231 cells decreased as arctigenin concentrations increased, with SK-BR-3 cell viability reduced by 62.1% and MDA-MB-231 cell viability reduced by 59.4% at 6.25 μM arctigenin (Fig. 1C, 1D).

Fig. 1. Effect of arctigenin on the viability of cancer cell lines. Cells were treated with an indicated dose of arctigenin for 24 hours and cell viability was measured by WST assay. (A) AGS. (B) MCF-7 (ER+, PR–, HER2–). (C) MDA-MB-231 (ER–, PR–, HER2–). (D) SK-BR-3 (ER–, PR–, HER2+). Results from duplicate experiments were analyzed by Student’s t-test (**P < 0.01 and ***P < 0.001). ER, estrogen receptor; PR, progesterone receptor; HER, human epidermal growth factor receptor 2.

2. Arctigenin-induced apoptosis in ER-negative SK-BR-3 cells

To determine whether arctigenin induces apoptosis, SK-BR-3 cells were treated with 0, 125, 250, and 500 nM arctigenin. Apoptosis was assessed using Annexin V-FITC/PI staining and flow cytometry, and PARP cleavage was evaluated by Western blot analysis. Flow cytometry revealed that the percentage of apoptotic cells increased from 4.14% to 6.34%, 7.92%, and 8.20% at 0, 125, 250, and 500 nM arctigenin, respectively (Fig. 2A, 2B). Western blot analysis demonstrated a decrease in full-length PARP and an increase in cleaved PARP with higher arctigenin concentrations (Fig. 2C, 2D). These findings indicate that arctigenin induces apoptosis in SK-BR-3 cells in a dose-dependent manner.

Fig. 2. Arctigenin induces apoptosis in SK-BR-3 cells. (A) SK-BR-3 cells were treated with an indicated dose of arctigenin for 24 hours, then stained with annexin V-FITC and PI, and subjected to flow cytometry. Stained cells were analyzed and illustrated on the quadrant by CellQuestPro software. (B) The percentage of apoptotic or live cells from flow cytometry results was analyzed and illustrated as a graph. (C) The cell lysates were subjected to Western blot to evaluate the levels of PARP, cleaved PARP, caspase 3, cleaved caspase 3, caspase 8, cleaved caspase 8, caspase 9, cleaved caspase 9, Survivin, and XIAP. GAPDH was used as an internal control. (D) Densities of the Western blot bands were illustrated as a graph and the results from triplicate experiments were analyzed by Student’s t-test (*P < 0.05, **P < 0.01, and ***P < 0.001).

3. Mechanisms of apoptosis induction by arctigenin in ER-negative SK-BR-3 cells

To further elucidate the mechanism of apoptosis induction by arctigenin, we analyzed the expression of caspases in SK-BR-3 cells treated with arctigenin. Western blot analysis showed a reduction in the levels of full-length caspases 3, 8, and 9, accompanied by an increase in their cleaved forms (Fig. 2C, 2D). These results suggest that arctigenin induces apoptosis in SK-BR-3 cells through a caspase-dependent mechanism involving both intrinsic (via caspase-9) and extrinsic (via caspase-8) pathways.

IAPs, such as XIAP and Survivin, inhibit apoptosis by directly binding to and inhibiting caspases, including caspases 3, 7, and 9. The expression levels of XIAP and Survivin in SK-BR-3 cells treated with arctigenin were assessed using Western blot analysis. The results indicate that arctigenin treatment led to a reduction in the levels of XIAP and Survivin, with a pronounced decrease observed at 500 nM arctigenin (Fig. 2C, 2D).

4. Increased Bax/Bcl-2 ratio in ER-negative SK-BR-3 cells treated with arctigenin

The Bcl-2 protein family regulates the intrinsic pathway of apoptosis, with Bax promoting and Bcl-2 inhibiting apoptosis. To evaluate the effect of arctigenin on these proteins, we measured the expression of Bax (pro-apoptotic) and Bcl-2 (anti-apoptotic) in SK-BR-3 cells by Western blot analysis and calculated the Bax/Bcl-2 ratio. Arctigenin treatment resulted in increased Bax expression and decreased Bcl-2 expression (Fig. 3A). The Bax/Bcl-2 ratio increased in a dose-dependent manner, reaching 4.7, 8.7, and 9.6 times that of untreated cells following treatment with 125, 250, and 500 nM arctigenin, respectively (Fig. 3B). These findings suggest that arctigenin-induced apoptosis is associated with the suppression of anti-apoptotic proteins (XIAP, Survivin, and Bcl-2) and the upregulation of the pro-apoptotic protein Bax in SK-BR-3 cells.

Fig. 3. Arctigenin increases the Bax/Bcl-2 ratio in SK-BR-3 cells. SK-BR-3 cells were treated with an indicated dose of arctigenin for 24 hours. The cell lysates were subjected to Western blot. (A) The level of Bax and Bcl-2. (B) Each band intensity from the data was normalized to GAPDH and the Bax/Bcl-2 ratio was calculated. Data were presented as mean ± standard error of the mean of three independent experiments and analyzed by Student’s t-test. ***P < 0.001 vs. non-treated control in SK-BR-3 cells.

5. Mechanisms of apoptosis induction by arctigenin in triple-negative MDA-MB-231 cells

The effect of arctigenin on apoptosis in MDA-MB-231 cells was evaluated by treating cells with 0, 125, 250, and 500 nM arctigenin for 24 hours, followed by flow cytometry using Annexin V-FITC/PI staining and Western blot analysis. Flow cytometry results demonstrated an increase in apoptosis in arctigenin-treated cells, with the apoptosis ratio rising from 7.13% in untreated cells to 8.98%, 10.86%, and 13.36% at 125, 250, and 500 nM arctigenin, respectively (Fig. 4A, 4B). Western blot analysis revealed a decrease in full-length PARP, caspase 3, and caspase 9, alongside an increase in cleaved PARP, cleaved caspase 3, and cleaved caspase 9 (Fig. 4C, 4D). Additionally, XIAP and Survivin levels were reduced following arctigenin treatment (Fig. 4C, 4D). The Bax/Bcl-2 ratio increased 2.2, 3.2, and 4.1 times in arctigenin-treated cells compared to untreated cells (Fig. 5A, 5B). These results indicate that arctigenin induces apoptosis in MDA-MB-231 cells primarily through the intrinsic apoptotic pathway.

Fig. 4. Arctigenin induces apoptosis in MDA-MB-231 cells. (A) MDA-MB-231 cells were treated with an indicated dose of arctigenin for 24 hours, then stained with annexin V-FITC and PI, and subjected to flow cytometry analysis. Stained cells were analyzed and illustrated on the quadrant by CellQuestPro software. (B) The percentage of apoptotic or live cells was analyzed and illustrated as a graph. (C) The cell lysates were subjected to Western blot to evaluate the levels of PARP, cleaved PARP, caspase 3, cleaved caspase 3, caspase 9, cleaved caspase 9, Survivin, and XIAP. GAPDH was used as an internal control. (D) Densities of the Western blot bands were illustrated as a graph and the results from triplicate experiments were analyzed by Student’s t-test (*P < 0.05, **P < 0.01, and ***P < 0.001).

Fig. 5. Arctigenin increases the Bax/Bcl-2 ratio in MDA-MB-231 cells. MDA-MB-231 cells were treated with an indicated dose of arctigenin for 24 hours. The cell lysates were subjected to Western blot. (A) The level of Bax and Bcl-2. (B) Each band intensity from the data was normalized to GAPDH and the Bax/Bcl-2 ratio was calculated. Data were presented as mean ± standard error of the mean of three independent experiments and analyzed by Student’s t-test. ***P < 0.001 vs. non-treated control in MDA-MB-231 cells.
DISCUSSION

In this study, we evaluated the inhibitory effects of arctigenin on three types of breast cancer cell lines: MCF-7 (ER+/HER2–/PR–), SK-BR-3 (ER–/HER2+/PR–) and MDA-MB-231 (ER–/HER2–/PR–). To evaluate the inhibitory effects of arctigenin on ER-negative breast cancer cells, ER-positive MCF-7 cells were used as a control. A gastric adenocarcinoma AGS cells were also used as a control.

Arctigenin significantly reduced the growth of ER-negative breast cancer cells. SK-BR-3 and MDA-MB-231 cells but did not affect AGS and MCF-7 cell viability. Specifically, arctigenin treatment activated caspases 3 and 9, induced PARP cleavage, and decreased the levels of anti-apoptotic proteins XIAP and Survivin in ER– SK-BR-3 and ER– MDA-MB-231 cells. Additionally, arctigenin treatment increased the Bax/Bcl-2 ratio in these cells, which likely contributed to the activation of caspase 9 and subsequent apoptosis through the intrinsic pathway. If apoptosis was induced in AGS or MCF-7 cells (control groups) by arctigenin treatment, the apoptotic extrinsic pathway would have been mainly activated (activation of Cas-7, -8, -3, and PARP). However, as shown in the WST assay results in result 1, AGS and MCF-7, which were used as controls, did not repeatedly show death in the concentration range set for arctigenin treatment. Therefore, we did not perform Western blotting on AGS and MCF-7. WST is a test that measures the color development of formazan reduced by succinate-tetrazolium reductase, a dehydrogenase present in the respiratory chain of mitochondria and active only in living cells.

Cell viability assays using the WST assay demonstrated that arctigenin did not significantly alter the viability of AGS cells at concentrations ranging from 0.39 μM to 50 μM (Fig. 1A). Susanti et al. (18) reported no cytotoxic effects on normal diploid fibroblasts (WI-38), human fibroblasts (KMST-6), normal embryo fibroblasts (OUMS-36), and normal mammary epithelial cells (H184B5F5/M10) under 50 μM of arctigenin treatment.

MCF-7 cells exhibited almost no reduction in viability at 50 μM arctigenin, while SK-BR-3 cells demonstrated a 22% and 62.1% decrease at the same concentrations, and MDA-MB-231 cells showed a 15.7% and 59.4% decrease in viability at 0.39 μM and 6.25 μM, respectively (Fig. 1B-1D). These findings are consistent with Hsieh et al. (19), who observed that arctigenin inhibited MDA-MB-231 cell growth but had minimal impact on MCF-7 cells at 20 μM for 48 hours. Conversely, Maxwell et al. (8) reported that arctigenin induced MCF-7 cell death through autophagy rather than apoptosis or cell cycle arrest, with a 30% reduction in cell viability at 100 μM for 24 hours. In our result, arctigenin showed decreased cell viability as 22% ± 2% at 100 μM for 24 hours (data not shown).

Feng et al. (5) identified IC50 values of 40 μM and 0.79 μM for MCF-7 and MDA-MB-231 cells, respectively, with arctigenin, correlating reduced MDA-MB-231 viability with apoptosis. Combined with previous studies, our results indicate that the inhibitory effect of arctigenin on MCF-7 cells is less pronounced compared to its effect on MDA-MB-231 and SK-BR-3 cells. This differential sensitivity may be due to the presence of ER in MCF-7 cells, which could reduce the inhibitory effects of arctigenin on cell proliferation compared to ER-negative breast cancer cells (19).

Exposure to cytotoxic agents can induce various cellular responses, including necrosis, cell cycle arrest, autophagy, and apoptosis. Previous studies have demonstrated that arctigenin triggers apoptosis and cell cycle arrest in diverse cancer cell lines (5-9). The current study specifically investigated apoptosis induced by arctigenin. Annexin V-FITC/PI staining and increased levels of cleaved PARP, as detected by Western blot analysis, confirmed that arctigenin induces apoptosis in SK-BR-3 and MDA-MB-231 cells (Fig. 2, 4). Furthermore, arctigenin activates caspases 3, 8, and 9 in SK-BR-3 cells (Fig. 2C, 2D) and caspases 3 and 9 in MDA-MB-231 cells (Fig. 4C, 4D). These results suggest that arctigenin induces apoptosis in SK-BR-3 breast cancer cells (ER–/HER2+/PR–) via both extrinsic and intrinsic pathways, while in MDA-MB-231 cells (ER–/HER2–/PR–), arctigenin induces apoptosis primarily through the intrinsic pathway.

Arctigenin treatment resulted in decreased expression of the anti-apoptotic proteins XIAP and Survivin in both SK-BR-3 and MDA-MB-231 cells (Fig. 2, 4). Survivin, which is regulated by the cell cycle and is expressed predominantly during the G2/M phase, is often overexpressed in cancers, correlating with chemotherapy resistance, increased tumor recurrence, and reduced patient survival. Thus, targeting Survivin represents a promising therapeutic approach (12).

The Bcl-2 family proteins are crucial in regulating the intrinsic apoptotic pathway. This family includes both pro-apoptotic proteins, such as Bax, and anti-apoptotic proteins, such as Bcl-2. Bax promotes the release of cytochrome c from the mitochondria, while Bcl-2 inhibits this release by interfering with Bax activity (11). Released cytochrome c binds to caspase-9, thereby initiating the intrinsic apoptosis pathway. The Bax/Bcl-2 ratio is critical in modulating cytochrome c release and mitochondrial membrane permeability. As shown in Fig. 3, 5, arctigenin increased Bax expression and decreased Bcl-2 expression, leading to enhanced mitochondrial membrane permeability, increased cytochrome c release, and activation of the intrinsic apoptotic pathway.

Based on these findings, arctigenin may be beneficial in the management of ER-negative and TNBCs. However, further in vivo studies are necessary to fully assess the anticancer efficacy of arctigenin.

Acknowledgement

None.

Conflict of interest

No potential conflict of interest relevant to this article was reported.

Funding

This work was supported by the National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIT) (No. RS-2023-00253761).

Authors’ contribution

Conceptualization: THY, SHK. Data curation: SHK, HJW. Formal analysis: SHK, HJW. Funding acquisition: SHK. Investigation: THY, HJW. Methodology: THY, HJW. Project administration: HJY, SHK. Resources: HJW, SHK. Software: HJW, THY. Supervision: SHK, HJW. Validation: SHK. Visualization: THY, HJW. Writing – original draft: THY. Writing – review and editing: SHK, HJW.

References
  1. Chan YS, Cheng LN, Wu JH, Chan E, Kwan YW, Lee SM, et al. A review of the pharmacological effects of Arctium lappa (burdock). Inflammopharmacology 2011;19:245-54.
    Pubmed CrossRef
  2. Kang HS, Lee JY, Kim CJ. Anti-inflammatory activity of arctigenin from Forsythiae Fructus. J Ethnopharmacol 2008;116:305-12.
    Pubmed CrossRef
  3. Pereira JV, Bergamo DC, Pereira JO, França Sde C, Pietro RC, Silva-Sousa YT. Antimicrobial activity of Arctium lappa constituents against microorganisms commonly found in endodontic infections. Braz Dent J 2005;16:192-6.
    Pubmed CrossRef
  4. Swarup V, Ghosh J, Mishra MK, Basu A. Novel strategy for treatment of Japanese encephalitis using arctigenin, a plant lignan. J Antimicrob Chemother 2008;61:679-88.
    Pubmed CrossRef
  5. Feng T, Cao W, Shen W, Zhang L, Gu X, Guo Y, et al. Arctigenin inhibits STAT3 and exhibits anticancer potential in human triple-negative breast cancer therapy. Oncotarget 2017;8:329-44.
    Pubmed KoreaMed CrossRef
  6. Jeong JB, Hong SC, Jeong HJ, Koo JS. Arctigenin induces cell cycle arrest by blocking the phosphorylation of Rb via the modulation of cell cycle regulatory proteins in human gastric cancer cells. Int Immunopharmacol 2011;11:1573-7.
    Pubmed CrossRef
  7. Maxwell T, Lee KS, Kim S, Nam KS. Arctigenin inhibits the activation of the mTOR pathway, resulting in autophagic cell death and decreased ER expression in ER-positive human breast cancer cells. Int J Oncol 2018;52:1339-49.
    Pubmed CrossRef
  8. Wang HQ, Jin JJ, Wang J. Arctigenin enhances chemosensitivity to cisplatin in human nonsmall lung cancer H460 cells through downregulation of survivin expression. J Biochem Mol Toxicol 2014;28:39-45.
    Pubmed CrossRef
  9. Yang S, Ma J, Xiao J, Lv X, Li X, Yang H, et al. Arctigenin anti-tumor activity in bladder cancer T24 cell line through induction of cell-cycle arrest and apoptosis. Anat Rec (Hoboken) 2012;295:1260-6.
    Pubmed CrossRef
  10. Wong RS. Apoptosis in cancer: from pathogenesis to treatment. J Exp Clin Cancer Res 2011;30:87.
    Pubmed KoreaMed CrossRef
  11. Tsujimoto Y, Shimizu S. Bcl-2 family: life-or-death switch. FEBS Lett 2000;466:6-10.
    Pubmed CrossRef
  12. Altieri DC. Survivin, versatile modulation of cell division and apoptosis in cancer. Oncogene 2003;22:8581-9.
    Pubmed CrossRef
  13. Eckelman BP, Salvesen GS, Scott FL. Human inhibitor of apoptosis proteins: why XIAP is the black sheep of the family. EMBO Rep 2006;7:988-94.
    Pubmed KoreaMed CrossRef
  14. Azamjah N, Soltan-Zadeh Y, Zayeri F. Global trend of breast cancer mortality rate: a 25-year study. Asian Pac J Cancer Prev 2019;20:2015-20.
    Pubmed KoreaMed CrossRef
  15. DeSantis CE, Ma J, Gaudet MM, Newman LA, Miller KD, Goding Sauer A, et al. Breast cancer statistics, 2019. CA Cancer J Clin 2019;69:438-51.
    Pubmed CrossRef
  16. Prat A, Pineda E, Adamo B, Galván P, Fernández A, Gaba L, et al. Clinical implications of the intrinsic molecular subtypes of breast cancer. Breast 2015;24 Suppl 2:S26-35.
    Pubmed CrossRef
  17. Romond EH, Perez EA, Bryant J, Suman VJ, Geyer CE Jr, Davidson NE, et al. Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer. N Engl J Med 2005;353:1673-84.
    Pubmed CrossRef
  18. Susanti S, Iwasaki H, Inafuku M, Taira N, Oku H. Mechanism of arctigenin-mediated specific cytotoxicity against human lung adenocarcinoma cell lines. Phytomedicine 2013;21:39-46.
    Pubmed CrossRef
  19. Hsieh CJ, Kuo PL, Hsu YC, Huang YF, Tsai EM, Hsu YL. Arctigenin, a dietary phytoestrogen, induces apoptosis of estrogen receptor-negative breast cancer cells through the ROS/p38 MAPK pathway and epigenetic regulation. Free Radic Biol Med 2014;67:159-70.
    Pubmed CrossRef